1
|
Eldehna WM, Habib YA, Mahmoud AE, Barghash MF, Elsayed ZM, Elsawi AE, Maklad RM, Rashed M, Khalil A, Hammad SF, Ali MM, El Kerdawy AM. Design, synthesis, and in silico insights of novel N'-(2-oxoindolin-3-ylidene)piperidine-4-carbohydrazide derivatives as VEGFR-2 inhibitors. Bioorg Chem 2024; 153:107829. [PMID: 39317037 DOI: 10.1016/j.bioorg.2024.107829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Vascular endothelial growth factor (VEGF) is a crucial key factor in breast tumorigenesis. VEGF plays an important role in angiogenesis, tumor proliferation, and metastasis. Herein, we report the design and synthesis of twenty-one novel piperidine/oxindole derivatives as potential VEGFR-2 inhibitors. The designed compound library aimed to occupy the binding site of VEGFR-2 in a similar binding pattern to that of the reference VEGFR-2 inhibitor Sorafenib. The synthesized compounds were biologically evaluated for their cytotoxic effects against two breast cancer cell lines (MCF-7 and MDA-MB-468). Compounds 12e and 6n were the most potent cytotoxic derivatives against the former and the latter cell lines, showing IC50 values of 8.00 and 0.60 µM, respectively. Furthermore, all the synthesized compounds were evaluated for their inhibitory activities towards VEGFR-2, with compound 12e showing the most potent activity with an IC50 value of 45.9 nM, surpassing the reference standard Sorafenib (IC50 = 48.6 nM). Additionally, compound 6n emerged as the top performer when tested with the other most promising compounds for their cytotoxic effects on HUVEC (IC50 = 28.77 nM). The designed library of compounds was subjected to molecular docking and molecular dynamic simulations, which revealed key binding interactions within the VEGFR-2 active site, including hydrogen bonding with Cys919, Glu885, and Asp1046 residues. Moreover, in silico predictions of physicochemical and pharmacokinetic properties for the target compounds indicated favorable drug-like characteristics.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt.
| | - Youmna A Habib
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Abeer E Mahmoud
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Mohamed F Barghash
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ahmed E Elsawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt
| | - Raed M Maklad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Amira Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Sherif F Hammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt; PharmD Program, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| | - Mamdouh M Ali
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, Lincolnshire, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. Box 11562 Cairo, Egypt
| |
Collapse
|
2
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
3
|
Lozon L, Ramadan WS, Kawaf RR, Al-Shihabi AM, El-Awady R. Decoding cell death signalling: Impact on the response of breast cancer cells to approved therapies. Life Sci 2024; 342:122525. [PMID: 38423171 DOI: 10.1016/j.lfs.2024.122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/04/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Breast cancer is a principal cause of cancer-related mortality in female worldwide. While many approved therapies have shown promising outcomes in treating breast cancer, understanding the intricate signalling pathways controlling cell death is crucial for optimizing the treatment outcome. A growing body of evidence has unveiled the aberrations in multiple cell death pathways across diverse cancer types, highlighting these pathways as appealing targets for therapeutic interventions. In this review, we provide a comprehensive overview of the current state of knowledge on the cell death signalling mechanisms with a particular focus on their impact on the response of breast cancer cells to approved therapies. Additionally, we discuss the potentials of combination therapies that exploit the synergy between approved drugs and therapeutic agents targeting modulators of cell death pathways.
Collapse
Affiliation(s)
- Lama Lozon
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rawan R Kawaf
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Aya M Al-Shihabi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
4
|
Wang K, Liao PY, Chang WC, Yang CR, Su YT, Wu PC, Wu YC, Hung YC, Akhtar N, Lai HC, Ma WL. Linoleate-pazopanib conjugation as active pharmacological ingredient to abolish hepatocellular carcinoma growth. Front Pharmacol 2024; 14:1281067. [PMID: 38293667 PMCID: PMC10824963 DOI: 10.3389/fphar.2023.1281067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Small molecule compounds targeting multiple kinases involved in neoangiogenesis have shown survival benefits in patients with unresectable hepatocellular carcinoma (HCC). Nonetheless, despite the beneficial effects of multikinase inhibitors (MKIs), a lack of boosting adjuvant limits their objective response rate. Lipid conjugates have been used to improve delivery efficacy or pharmaceutical benefits for decades. However, the feasibility of utilizing lipid-drug conjugates (LDCs) in HCC regimens remains untested. In this study, oral feeding of linoleate-fluorescein isothiocyanate conjugates showed that the compound was well distributed in a spontaneous HCC mouse model. Therefore, a rationale design was developed for chemically synthesizing a linoleate-pazopanib conjugate (LAPC). The LAPC showed a significantly improved cytotoxicity compared to the parental drug pazopanib. Pazopanib's angiogenic suppressing signals were not observed in LAPC-treated HCC cells, potentially suggesting an altered mechanism of action (MOA). In an efficacy trial comparing placebo, oral pazopanib, and LAPC treatments in the hepatitis B virus transgene-related spontaneous HCC mouse model (HBVtg-HCC), the LAPC treatment demonstrated superior tumor ablating capacity in comparison to both placebo and pazopanib treatments, without any discernible systemic toxicity. The LAPC exposure is associated with an apoptosis marker (Terminal deoxynucleotidyl transferase dUTP nick end labeling [TUNEL]) and an enhanced ferroptosis (glutathione peroxidase 4 [GPX4]) potential in HBVtg-HCC tumors. Therefore, the LAPC showed excellent HCC ablative efficacy with altered MOA. The molecular mechanisms of the LAPC and LDCs for HCC therapeutics are of great academic interest. Further comprehensive preclinical trials (e.g., chemical-manufacture-control, toxicity, distribution, and pharmacokinetics/pharmacodynamics) are expected.
Collapse
Affiliation(s)
- Ke Wang
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Yin Liao
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chang
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Cian-Ru Yang
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Ting Su
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Ching Wu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, Taiwan Innovation Center of Medical Devices and Technology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yao-Ching Hung
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Asia University Hospital, Taichung, Taiwan
| | - Najim Akhtar
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lung Ma
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Huang W, Wang C, Shen Y, Chen Q, Huang Z, Liu J, Lin X, Wang L, Wu F, Chen X, Li N, Hong Y, Chen M, Li J, Huang C. A real-world study of the effectiveness and safety of apatinib-based regimens in metastatic triple-negative breast cancer. BMC Cancer 2024; 24:39. [PMID: 38182995 PMCID: PMC10768098 DOI: 10.1186/s12885-023-11790-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/22/2023] [Indexed: 01/07/2024] Open
Abstract
PURPOSE This investigation sought to examine the efficacy and safety of low-dose apatinib used alongside chemotherapy in the clinical management of patients with metastatic triple-negative breast cancer (TNBC) within a real-world setting, whilst comparing the outcomes with those treated solely with chemotherapy. METHODS This case series study analyzed clinical data and treatment outcomes of 163 patients with metastatic TNBC who underwent rescue treatment at the Medical Oncology Department of Clinical Oncology, Fujian Cancer Hospital, School of Fujian Medical University, China, between October 2011 and January 2023. All the patients underwent rescue treatment with either chemotherapy alone or apatinib (250 mg/day) combined with chemotherapy. The study's primary outcome was progression-free survival (PFS), whereas the secondary outcomes included overall survival (OS), objective response rate (ORR), disease control rate (DCR), and safety profiles. RESULTS The study was designed to compare two groups [1]. Out of the 163 TNBC patients who participated in the study, 107 individuals (65.6%) received treatment based on chemotherapy, whereas 56 patients (34.4%) were given treatment based on a combination of low-dose apatinib (250 mg/day) and other treatments, including chemotherapy. After propensity score matching (PSM), the objective response rate (ORR) and disease control rate (DCR) of patients with advanced triple-negative breast cancer (TNBC) who received apatinib-based treatment were 50.0 and 90.0%, respectively, while they were 6.7 and 20.0%, respectively, for the chemotherapy-based group (P < 0.001). The group that received apatinib-based treatment showed superior results in both PFS and OS compared to the group that received chemotherapy. The median PFS and OS for the apatinib-based group were 7.8 and 20.3 months, respectively, while they were only 2.2 months and 9.0 months, respectively, for the chemotherapy-based group (P < 0.001) [2]. Patients who were administered combo therapies, including PD-1 inhibitors, were excluded. In total, 97 patients received chemotherapy alone, while 34 patients were treated with apatinib in combination with chemotherapy. After propensity score matching (PSM), the ORR and DCR for the total group who received combo therapies were 44.4 and 81.5%, respectively, while they were 11.1 and 22.2%, respectively, for the chemotherapy alone group (P < 0.001). The group receiving both apatinib and chemotherapy displayed notable advantages over the group solely receiving chemotherapy in regards to PFS and OS for the entirety of the population. The PFS was found to be 7.8 months in comparison to 2.1 months (P < 0.001) and the OS was 21.1 months in contrast to 9.0 months (P < 0.001). Apatinib combined with chemotherapy induced grade 3/4 hematological toxicities, including neutropenia (8.8%) and thrombocytopenia (2.9%). Additionally, non-hematological toxicities were commonly observed, such as Hand-foot syndrome (35.3%), proteinuria (26.5%), hypertension (61.8%), higher alanine aminotransferase levels (26.5%), and fatigue (35.3%). The most frequent non-hematological grade 3/4 toxicities were Hand-foot syndrome (2.9%) and hypertension (5.9%). The study did not report any fatal adverse effects. CONCLUSIONS The combination of low-dose apatinib with chemotherapy has proven to be more effective than chemotherapy alone in treating metastatic triple-negative breast cancer (TNBC). Additionally, the occurrence of grade 3/4 non-hematologic toxicities was significantly lower compared to the recommended dose of apatinib.
Collapse
Affiliation(s)
- Weiwei Huang
- Department of Medical Oncology, Fujian Medical University Union Hospital, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian province, 350001, China
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospotial, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Chenxi Wang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province, 350117, PR China
| | - Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province, 350117, PR China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province, 350117, PR China
| | - Zhijian Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Jian Liu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Xiaoyan Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian province, 350001, China.
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospotial, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China.
| | - Lili Wang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Fan Wu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Xinhua Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Nani Li
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Yi Hong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Mulan Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Jieyu Li
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| | - Chuanzhong Huang
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian province, 350014, China
| |
Collapse
|
6
|
Tamimi A, Tamimi A, Sorkheh F, Asl SM, Ghafari A, Karimi AG, Erabi G, Pourmontaseri H, Deravi N. Monoclonal antibodies for the treatment of squamous cell carcinoma: A literature review. Cancer Rep (Hoboken) 2023; 6:e1802. [PMID: 37042307 PMCID: PMC10172176 DOI: 10.1002/cnr2.1802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 02/12/2023] [Accepted: 02/24/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Squamous cell carcinoma (SCC) is a relatively common and heterogenous malignancy of different organs, such as the skin, esophagus, and lungs. Although most cases experience good survival with surgical methods, management of advanced types of the disease remains challenging. Several modalities, including different chemotherapy regimens and immunotherapies, have been investigated in this matter, among which Monoclonal antibodies (Mabs) are one of the most promising ones. Since the development of Mabs, they have been widely used to treat different diseases. Mabs have shown significant efficacy with high specificity along with acceptable safety, which makes them a favorable option in cancer therapy. In this article, we aimed to review the different aspects of using Mabs in SCC therapy. RECENT FINDINGS We found that treating with different Mabs has shown excellent efficacy accompanied by acceptable safety in treating SCC of different organs. Therefore, Mabs are considered great options in the treatment of SCC, especially in advanced cases. Overall, two highly potent types of Mabs in SCC therapy are anti-EGFR Mabs and checkpoint inhibitors, especially Cetuximab, Nimotuzumab, and PD-1 inhibitors. Bevacizumab is also a promising option as adjuvant therapy to other modalities. CONCLUSION Although some Mabs have shown promising outcomes in SCC therapy, their application as a part of cancer treatment depends on further investigations regarding cost-effectiveness and predictors of response. FDA has approved several Mabs in SCC therapies, and Mabs may have a crucial role in this era in the near future, especially in treating head and neck and esophageal SCC and metastatic lung cancer.
Collapse
Affiliation(s)
- Amirhossein Tamimi
- Student Research Committee, School of MedicineGuilan University of Medical SciencesRashtIran
| | - Atena Tamimi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fatemeh Sorkheh
- Student Research CommitteeBabol University of Medical SciencesBabolIran
| | - Saba Mardekatani Asl
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Arezoo Ghafari
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | | - Gisou Erabi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | - Hossein Pourmontaseri
- Student Research CommitteeFasa University of Medical SciencesFasaIran
- Bitab knowledge EnterpriseFasa University of Medical SciencesFasaIran
| | - Niloofar Deravi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
7
|
Dzhuzha A, Gandalipov E, Korzhikov-Vlakh V, Katernyuk E, Zakharova N, Silonov S, Tennikova T, Korzhikova-Vlakh E. Amphiphilic Polypeptides Obtained by Post-Polymerization Modification of Poly-l-Lysine as Systems for Combined Delivery of Paclitaxel and siRNA. Pharmaceutics 2023; 15:pharmaceutics15041308. [PMID: 37111793 PMCID: PMC10143851 DOI: 10.3390/pharmaceutics15041308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
The development of effective anti-cancer therapeutics remains one of the current pharmaceutical challenges. The joint delivery of chemotherapeutic agents and biopharmaceuticals is a cutting-edge approach to creating therapeutic agents of enhanced efficacy. In this study, amphiphilic polypeptide delivery systems capable of loading both hydrophobic drug and small interfering RNA (siRNA) were developed. The synthesis of amphiphilic polypeptides included two steps: (i) synthesis of poly-αl-lysine by ring-opening polymerization and (ii) its post-polymerization modification with hydrophobic l-amino acid and l-arginine/l-histidine. The obtained polymers were used for the preparation of single and dual delivery systems of PTX and short double-stranded nucleic acid. The obtained double component systems were quite compact and had a hydrodynamic diameter in the range of 90-200 nm depending on the polypeptide. The release of PTX from the formulations was studied, and the release profiles were approximated using a number of mathematical dissolution models to establish the most probable release mechanism. A determination of the cytotoxicity in normal (HEK 293T) and cancer (HeLa and A549) cells revealed the higher toxicity of the polypeptide particles to cancer cells. The separate evaluation of the biological activity of PTX and anti-GFP siRNA formulations testified the inhibitory efficiency of PTX formulations based on all polypeptides (IC50 4.5-6.2 ng/mL), while gene silencing was effective only for the Tyr-Arg-containing polypeptide (56-70% GFP knockdown).
Collapse
Affiliation(s)
- Apollinariia Dzhuzha
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Erik Gandalipov
- International Institute of Solution Chemistry and Advanced Materials Technologies, ITMO University, Lomonosov Street 9, St. Petersburg 191002, Russia
| | - Viktor Korzhikov-Vlakh
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
| | - Elena Katernyuk
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Natalia Zakharova
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Sergey Silonov
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Cytology, Russian Academy of Sciences, Tihkorezky Pr. 4, St. Petersburg 194064, Russia
| | - Tatiana Tennikova
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| |
Collapse
|
8
|
Jing H, Wu X, Xiang M, Wang C, Novakovic VA, Shi J. Microparticle Phosphatidylserine Mediates Coagulation: Involvement in Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:cancers15071957. [PMID: 37046617 PMCID: PMC10093313 DOI: 10.3390/cancers15071957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Tumor progression and cancer metastasis has been linked to the release of microparticles (MPs), which are shed upon cell activation or apoptosis and display parental cell antigens, phospholipids such as phosphatidylserine (PS), and nucleic acids on their external surfaces. In this review, we highlight the biogenesis of MPs as well as the pathophysiological processes of PS externalization and its involvement in coagulation activation. We review the available evidence, suggesting that coagulation factors (mainly tissue factor, thrombin, and fibrin) assist in multiple steps of tumor dissemination, including epithelial-mesenchymal transition, extracellular matrix remodeling, immune escape, and tumor angiogenesis to support the formation of the pre-metastatic niche. Platelets are not just bystander cells in circulation but are functional players in primary tumor growth and metastasis. Tumor-induced platelet aggregation protects circulating tumor cells (CTCs) from the blood flow shear forces and immune cell attack while also promoting the binding of CTCs to endothelial cells and extravasation, which activates tumor invasion and sustains metastasis. Finally, in terms of therapy, lactadherin can inhibit coagulation by competing effectively with coagulation factors for PS binding sites and may similarly delay tumor progression. Furthermore, we also investigate the therapeutic potential of coagulation factor inhibitors within the context of cancer treatment. The development of multiple therapies targeting platelet activation and platelet-tumor cell interactions may not only reduce the lethal consequences of thrombosis but also impede tumor growth and spread.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Chengyue Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02132, USA
| |
Collapse
|
9
|
Bai JW, Qiu SQ, Zhang GJ. Molecular and functional imaging in cancer-targeted therapy: current applications and future directions. Signal Transduct Target Ther 2023; 8:89. [PMID: 36849435 PMCID: PMC9971190 DOI: 10.1038/s41392-023-01366-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 03/01/2023] Open
Abstract
Targeted anticancer drugs block cancer cell growth by interfering with specific signaling pathways vital to carcinogenesis and tumor growth rather than harming all rapidly dividing cells as in cytotoxic chemotherapy. The Response Evaluation Criteria in Solid Tumor (RECIST) system has been used to assess tumor response to therapy via changes in the size of target lesions as measured by calipers, conventional anatomically based imaging modalities such as computed tomography (CT), and magnetic resonance imaging (MRI), and other imaging methods. However, RECIST is sometimes inaccurate in assessing the efficacy of targeted therapy drugs because of the poor correlation between tumor size and treatment-induced tumor necrosis or shrinkage. This approach might also result in delayed identification of response when the therapy does confer a reduction in tumor size. Innovative molecular imaging techniques have rapidly gained importance in the dawning era of targeted therapy as they can visualize, characterize, and quantify biological processes at the cellular, subcellular, or even molecular level rather than at the anatomical level. This review summarizes different targeted cell signaling pathways, various molecular imaging techniques, and developed probes. Moreover, the application of molecular imaging for evaluating treatment response and related clinical outcome is also systematically outlined. In the future, more attention should be paid to promoting the clinical translation of molecular imaging in evaluating the sensitivity to targeted therapy with biocompatible probes. In particular, multimodal imaging technologies incorporating advanced artificial intelligence should be developed to comprehensively and accurately assess cancer-targeted therapy, in addition to RECIST-based methods.
Collapse
Affiliation(s)
- Jing-Wen Bai
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Medical Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Clinical Research Center, Shantou Central Hospital, 515041, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, 515041, Shantou, China
| | - Guo-Jun Zhang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
| |
Collapse
|
10
|
Maqsood Q, Sumrin A, Iqbal M, Hussain N, Mahnoor M, Zafar Saleem M, Perveen R. A Winning New Combination? Toward Clinical Application in Oncology. Cancer Control 2023; 30:10732748231175240. [PMID: 37166227 PMCID: PMC10184224 DOI: 10.1177/10732748231175240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/04/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023] Open
Abstract
Immunotherapy has substantial attention in oncology due to the success of CTLA-4 and PD-1 inhibitors in the treatment of melanoma, lung cancer, head and neck cancer, renal cell carcinoma, and Hodgkin's lymphoma. A deeper understanding of interaction of tumor with its environment and the immune system provides best guide for oncology research. Recent studies in oncology have explained how a tumor alters antigen presentation, avoids detection, and activation of the host immune system to live and develop. Understanding the connections between the tumor and the immune system has resulted in several innovative therapy options. The extensive field of gene therapy has provided a number of cutting-edge medicines that are expected to play an important role in lowering cancer-related mortality. This article explains the history, important breakthroughs, and future prospects for three separate gene therapy treatment modalities: immunotherapy, oncolytic virotherapy, and gene transfer. Immunotherapies have completely changed how cancer is treated, especially for individuals whose condition was previously thought to be incurable. Examples include ACT (adoptive cell therapy) and ICB (immune checkpoint blockade). This review article will discuss the relationship between the immune response to cancer and the mechanisms of immunotherapy resistance. It will cover combination drugs authorized by the US Food and Drug Administration and provide a thorough overview of how these drugs are doing clinically right now. Cytokines, vaccines, and other soluble immunoregulatory agents, innate immune modifiers, ACT, virotherapy, and other treatment modalities will all be covered in detail.
Collapse
Affiliation(s)
- Quratulain Maqsood
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| | - Aleena Sumrin
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| | - Maryam Iqbal
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| | - Nazim Hussain
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| | - Muhammada Mahnoor
- Department of Rehabilitation Sciences, Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | - Muhammad Zafar Saleem
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| | - Rukhsana Perveen
- Centre for Applied Molecular Biology, University of the Punjab Quaid-i-Azam Campus, Lahore, Pakistan
| |
Collapse
|
11
|
Yang C, Zhang J, Zhang Y, Ji F, Chen Y, Zhu T, Zhang L, Gao H, Yang M, Li J, Cheng M, Wang K. Low-dose apatinib combined with neoadjuvant chemotherapy in the treatment of early-stage triple-negative breast cancer (LANCET): a single-center, single-arm, phase II trial. Ther Adv Med Oncol 2022; 14:17588359221118053. [PMID: 35983024 PMCID: PMC9379563 DOI: 10.1177/17588359221118053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Antiangiogenic therapy combined with chemotherapy could improve pathological complete response (pCR) for breast cancer. Apatinib is an oral tyrosine kinase inhibitor that selectively inhibits vascular endothelial growth factor receptor 2. We assessed the efficacy and safety of apatinib combined with standard neoadjuvant chemotherapy in patients with triple-negative breast cancer (TNBC). Materials and methods: This single-arm, phase II study enrolled patients aged 18–70 years with previously untreated stage IIA-IIIB TNBC. Patients received oral apatinib at a dose of 250 mg once daily and intravenously docetaxel every 3 weeks for four cycles, followed by epirubicin plus cyclophosphamide every 3 weeks for four cycles. The primary endpoint was the pCR rate in the breast and lymph nodes. Secondary endpoints included objective response rate, event-free survival (EFS), overall survival (OS), and safety. Results: In all, 31 patients were enrolled, and the median follow-up time was 22.9 months (range: 10.1–41.6 months). The pCRs in both breast and lymph nodes were achieved in 17 [54.8%; 95% confidence interval (CI): 36.0–72.7] of 31 patients. Objective responses were achieved in 29 patients (93.5%; 95% CI: 78.6–99.2), and disease control was achieved in 31 patients (100%; 95% CI: 88.8–100.0). The 2-year EFS and 2-year OS were 90.9% and 94.4%, respectively. The five most common treatment-related adverse events were fatigue (51%), hypertension (41%), anorexia (39%), hand–foot syndrome (35%), and diarrhea (32%). Few grade 3 or more adverse events were observed. Conclusion: The combination of apatinib with docetaxel followed by epirubicin plus cyclophosphamide showed excellent efficacy and manageable toxicities; and further randomized controlled phase III trials are warranted. Trial registration: This trial was registered with ClinicalTrials.gov (NCT03243838) on 5 August 2017.
Collapse
Affiliation(s)
- Ciqiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junsheng Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fei Ji
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yitian Chen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liulu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hongfei Gao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Mei Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jieqing Li
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Minyi Cheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 123 Huifu West Road, Guangzhou, 510080, China
| |
Collapse
|
12
|
Ren Y, Pan F, Kan X, Wang J, Han P, Yan J, Li L, Sun P, Liu CY, Bao Q, Yang L, Zheng C. Multimodal Imaging Response after the Singular or Combination Treatments of Vascular Endothelial Growth Factor Inhibitor and Immune Checkpoint Inhibitor. Mol Pharm 2022; 19:3664-3672. [PMID: 35976154 DOI: 10.1021/acs.molpharmaceut.2c00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study aims to dynamically assess tumor changes after variable treatments with vascular endothelial growth factor (VEGF) inhibitor and/or immune checkpoint inhibitor (ICI) using multimodal imaging of MRI and 18F-FDG PET/CT in a hepatocellular carcinoma (HCC) mice model. Based on different treatments, 24 mice were randomly divided into four groups: control (isotype-matched IgG antibody 10 mg/kg), VEGF inhibitor (sorafenib 50 mg/kg), ICI (anti-PD-L1 antibody 10 mg/kg), and combination groups (sorafenib 50 mg/kg + anti-PD-L1 antibody 10 mg/kg). Quantitative imaging assessments, including volume transfer constant (Ktrans), apparent diffusion coefficient (ADC), lactate/choline ratio, and the maximum standardized 18F-FDG uptake value ratio of tumor to muscle (SUVtumor/SUVmuscle ratio), were acquired at different time points (before treatment and 7, 14, and 21 days after treatment). Quantitative data were presented as the mean ± standard errors and two-way repeated-measure ANOVA tests were performed for intergroup and intertime point comparisons. After 21 days from the initiation of therapies, combination group showed the lowest tumor volume and weight, followed by ICI, VEGF inhibitor, and control group, with no significance between the VEGF inhibitor and control groups. In addition, Ktrans values significantly decreased, and the lactate/choline ratio and SUVtumor/SUVmuscle ratio were significantly elevated in the VEGF inhibitor group. ADC significantly increased in the ICI and combination groups, with no significant differences in ADC observed between the control and VEGF inhibitor groups, which showed a similar dynamic change to the tumor volume. Furthermore, Ktrans, lactate/choline ratio, and ADC were significantly correlated with CD31+ area, hypoxyprobe+ area, and apoptosis, respectively. Our results suggest that the singular treatment and combination of the VEGF inhibitor and ICI treatments for HCC present different multimodal imaging changes in accordance with the specific histopathological features. These findings might facilitate the formulation of better treatment response criteria; besides, we find ADC is probably an indicator easily to obtain for treatment response evaluation.
Collapse
Affiliation(s)
- Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jiazheng Wang
- Clinical & Technical Solutions, Philips Healthcare, Beijing 100600, China
| | - Ping Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jingjie Yan
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathmatics, Innovation Academy for Precision Measurement Science and Technology, Wuhan 430071, China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lingli Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Peng Sun
- Clinical & Technical Solutions, Philips Healthcare, Beijing 100600, China
| | - Chao-Yang Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathmatics, Innovation Academy for Precision Measurement Science and Technology, Wuhan 430071, China
| | - Qingjia Bao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathmatics, Innovation Academy for Precision Measurement Science and Technology, Wuhan 430071, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
13
|
Wang Y, Song W, Zhou S, Chang S, Chang J, Tian J, Zhang L, Li J, Che G. The genomic and transcriptome characteristics of lung adenocarcinoma patients with previous breast cancer. BMC Cancer 2022; 22:618. [PMID: 35668376 PMCID: PMC9171992 DOI: 10.1186/s12885-022-09727-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Breast cancer and lung cancer are the top two malignancies in the female population and the number of patients with breast cancer and subsequent primary lung cancer has increased significantly in recent years. However, the unique molecular characteristics of this group of patients remains unclear. PURPOSE To identify the genomic and transcriptome characteristics of primary lung adenocarcinoma patients with previous breast cancer by comparison with single primary lung adenocarcinoma (SPLA) patients. METHODS The tumor and normal pulmonary tissue specimens of ten primary pulmonary adenocarcinoma patients with previous breast cancer (multiple primary cancer, MPC) and ten SPLA patients were prospectively collected. The whole exome sequencing (WES) and RNA sequencing (RNA-seq) were performed to analyze the gene mutation and expression differences between MPC and SPC patients. RESULTS The results of WES indicated that the mutations of TRIM73, DLX6 and CNGB1 only existed in MPC patients. The results of RNA-seq manifested the occurrence of second primary lung adenocarcinoma in breast cancer patients was closely associated with cytokine-cytokine receptor action, autophagy, PI3L-Akt, cAMP and calcium ion signaling pathways. Besides, the expression levels of FGF10 and VEGFA genes were significantly increased in MPC patients. CONCLUSION The occurrence of second primary lung adenocarcinoma may be related to the cytokine-cytokine receptor action, autophagy, PI3L-Akt, cAMP and calcium ion signaling pathways. Furthermore, the mutations of TRIM73, DLX6 and CNGB1 and high expression of FGF10 and VEGFA might play an important role in the development of lung adenocarcinoma in breast cancer patients. However, more in-depth investigations are needed to verify above findings.
Collapse
Affiliation(s)
- Yan Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Wenpeng Song
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Sicheng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Shuai Chang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Junke Chang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Liming Zhang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Jue Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Guoxuexiang No. 37, Chengdu, 610041, China.
| |
Collapse
|
14
|
Torres-Vergara P, Troncoso F, Acurio J, Kupka E, Bergman L, Wikström AK, Escudero C. Dysregulation of vascular endothelial growth factor receptor 2 phosphorylation is associated with disruption of the blood-brain barrier and brain endothelial cell apoptosis induced by plasma from women with preeclampsia. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166451. [DOI: 10.1016/j.bbadis.2022.166451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/27/2022]
|
15
|
Zhou C, O'Connor J, Backen A, Valle JW, Bridgewater J, Dive C, Jayson GC. Plasma Tie2 trajectories identify vascular response criteria for VEGF inhibitors across advanced biliary tract, colorectal and ovarian cancers. ESMO Open 2022; 7:100417. [PMID: 35279528 PMCID: PMC9058891 DOI: 10.1016/j.esmoop.2022.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor inhibitors (VEGFi) are compromised by a lack of validated biomarkers. Previously we showed that changes in the concentration of plasma Tie2 (pTie2) was a response biomarker for bevacizumab. Here, we investigated whether pTie2 can predict response and progression cross-tumour for generic VEGFi treatment. PATIENTS AND METHODS Patients (n = 124) with advanced biliary tract cancer (ABC) received cisplatin/gemcitabine with cediranib or placebo (ABC-03 trial). Concentrations of pTie2 were measured longitudinally from before treatment until disease progression. Data from patients with ovarian cancer (n = 92, ICON7 trial) and patients with colorectal cancer (CRC) (n = 70, Travastin trial) were also included. RESULTS Cediranib-treated ABC patients were deconvoluted into distinct groups where in one group pTie2 trajectories resembled those seen in placebo-treated patients and in another pTie2 significantly reduced (t-test P = 2.7 × 10-14). Using the 95% confidence interval for these two groups, we defined a vascular complete response (vCR) as a 24% reduction in pTie2 within 9 weeks; vascular no response (vNR) as a 7% increase in pTie2, and a vascular partial response (between these limits). vCR cediranib-treated patients had significantly improved progression-free survival (8.8 versus 7.5 months, restricted mean ratio 0.73, P = 0.012) and overall survival (18.8 versus 12.1 months, hazard ratio 0.49, P = 0.02). By integrating data across ovarian cancer, CRC and ABC, we show that (i) patients with vNR do not benefit from VEGFi and (ii) Tie2-defined vascular progression occurs sufficiently in advance of radiological progressive disease that changes in treatment could be offered to prevent clinical deterioration. CONCLUSION pTie2 is the first cross-tumour, generic VEGFi, vascular response biomarker to guide optimum use of VEGFi in clinical practice.
Collapse
Affiliation(s)
- C Zhou
- CRUK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - J O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - A Backen
- Division of Cancer Sciences, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK
| | - J W Valle
- The Christie NHS Foundation Trust, Manchester, UK
| | - J Bridgewater
- University College Hospital Macmillan Cancer Centre, Huntley Street, London, UK
| | - C Dive
- CRUK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - G C Jayson
- Division of Cancer Sciences, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
16
|
Hofmann E, Eggers B, Heim N, Kramer FJ, Nokhbehsaim M, Götz W. Bevacizumab and sunitinib mediate osteogenic and pro-inflammatory molecular changes in primary human alveolar osteoblasts in vitro. Odontology 2022; 110:634-647. [PMID: 35171372 PMCID: PMC9463285 DOI: 10.1007/s10266-022-00691-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/04/2022] [Indexed: 12/03/2022]
Abstract
Antiangiogenic medications target the de novo blood vessel formation in tumorigenesis. However, these novel drugs have been linked to the onset of medication-related osteonecrosis of the jaw (MRONJ). The aim of this in vitro study was to examine the effects of the vascular endothelial growth factor A (VEGFA) antibody bevacizumab (BEV) and the receptor tyrosine kinase inhibitor (RTKI) sunitinib (SUN) on primary human osteoblasts derived from the alveolar bone. Primary human alveolar osteoblasts (HAOBs) were treated with BEV or SUN for 48 h. Cellular metabolic activity was examined by XTT assay. Differentially regulated genes were identified by screening of 22 selected osteogenic and angiogenic markers by quantitative real-time reverse transcriptase polymerase chain reaction (qRT2-PCR). Protein levels of alkaline phosphatase (ALP), collagen type 1, α1 (COL1A1) and secreted protein acidic and cysteine rich (SPARC) were examined by enzyme-linked immunoassay (ELISA). Treatment with BEV and SUN did not exhibit direct cytotoxic effects in HAOBs as confirmed by XTT assay. Of the 22 genes examined by qRT2-PCR, four genes were significantly regulated after BEV treatment and eight genes in the SUN group as compared to the control group. Gene expression levels of ALPL, COL1A1 and SPARC were significantly downregulated by both drugs. Further analysis by ELISA indicated the downregulation of protein levels of ALP, COL1A1 and SPARC in the BEV and SUN groups. The effects of BEV and SUN in HAOBs may be mediated by alterations to osteogenic and catabolic markers. Therapeutic or preventive strategies in MRONJ may address drug-induced depression of osteoblast differentiation.
Collapse
Affiliation(s)
- Elena Hofmann
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Welschnonnenstr 17, 53111, Bonn, Germany.
- Department of Oral and Maxillofacial Surgery, Charité- Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Benedikt Eggers
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - Nils Heim
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - Franz-Josef Kramer
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Welschnonnenstr 17, 53111, Bonn, Germany
| | - Marjan Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, University Hospital Bonn, 53111, Bonn, Germany
| | - Werner Götz
- Department of Orthodontics, University Hospital Bonn, 53111, Bonn, Germany
| |
Collapse
|
17
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
18
|
Yadav K, Lim J, Choo J, Ow SGW, Wong A, Lee M, Chan CW, Hartman M, Lim SE, Ngoi N, Tang SW, Ang Y, Chan G, Chong WQ, Tan HL, Tan SH, Goh BC, Lee SC. Immunohistochemistry study of tumor vascular normalization and anti-angiogenic effects of sunitinib versus bevacizumab prior to dose-dense doxorubicin/cyclophosphamide chemotherapy in HER2-negative breast cancer. Breast Cancer Res Treat 2021; 192:131-142. [PMID: 34928481 PMCID: PMC8841320 DOI: 10.1007/s10549-021-06470-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022]
Abstract
Purpose Tumor angiogenesis controlled predominantly by vascular endothelial growth factor and its receptor (VEGF-VEGFR) interaction plays a key role in the growth and propagation of cancer cells. However, the newly formed network of blood vessels is disorganized and leaky. Pre-treatment with anti-angiogenic agents can “normalize” the tumor vasculature allowing effective intra-tumoral delivery of standard chemotherapy. Immunohistochemistry (IHC) analysis was applied to investigate and compare the vascular normalization and anti-angiogenic effects of two commonly used anti-angiogenic agents, Sunitinib and Bevacizumab, administered prior to chemotherapy in HER2-negative breast cancer patients. Methods This prospective clinical trial enrolled 38 patients into a sunitinib cohort and 24 into a bevacizumab cohort. All received 4 cycles of doxorubicin/cyclophosphamide chemotherapy and pre-treatment with either sunitinib or bevacizumab. Tumor biopsies were obtained at baseline, after cycle 1 (C1) and cycle 4 (C4) of chemotherapy. IHC was performed to assess the tumor vascular normalization index (VNI), lymphatic vessel density (LVD), Ki67 proliferation index and expression of tumor VEGFR2. Results In comparison to Bevacizumab, Sunitinib led to a significant increase in VNI post-C1 and C4 (p < 0.001 and 0.001) along with decrease in LVD post-C1 (p = 0.017). Both drugs when combined with chemotherapy resulted in significant decline in tumor proliferation after C1 and C4 (baseline vs post-C4 Ki67 index p = 0.006 for Sunitinib vs p = 0.021 for Bevacizumab). Bevacizumab resulted in a significant decrease in VEGFR2 expression post-C1 (p = 0.004). Conclusion Sunitinib, in comparison to Bevacizumab showed a greater effect on tumor vessel modulation and lymphangiogenesis suggesting that its administration prior to chemotherapy might result in improved drug delivery. Trial registry ClinicalTrials.gov: NCT02790580 (first posted June 6, 2016).
Collapse
Affiliation(s)
- Kritika Yadav
- Department of Pathology, Dr. D Y Patil Medical College, Navi Mumbai, India
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Joline Lim
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Joan Choo
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Samuel Guan Wei Ow
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Andrea Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Matilda Lee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Ching Wan Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Mikael Hartman
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Siew Eng Lim
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Natalie Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Siau Wei Tang
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Yvonne Ang
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Gloria Chan
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Wan Qin Chong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Hon Lyn Tan
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Sing Huang Tan
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Boon Cher Goh
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Soo Chin Lee
- Cancer Science Institute, National University of Singapore, Singapore, Singapore.
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore.
| |
Collapse
|
19
|
López-Vega JM, Álvarez I, Antón A, Illarramendi JJ, Llombart A, Boni V, García-Velloso MJ, Martí-Climent JM, Pina L, García-Foncillas J. Early Imaging and Molecular Changes with Neoadjuvant Bevacizumab in Stage II/III Breast Cancer. Cancers (Basel) 2021; 13:3511. [PMID: 34298725 PMCID: PMC8307791 DOI: 10.3390/cancers13143511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 11/24/2022] Open
Abstract
This prospective, phase II study evaluated novel biomarkers as predictors of response to bevacizumab in patients with breast cancer (BC), using serial imaging methods and gene expression analysis. Patients with primary stage II/III BC received bevacizumab 15 mg/kg (cycle 1; C1), then four cycles of neoadjuvant docetaxel doxorubicin, and bevacizumab every 3 weeks (C2-C5). Tumour proliferation and hypoxic status were evaluated using 18F-fluoro-3'-deoxy-3'-L-fluorothymidine (FLT)- and 18F-fluoromisonidazole (FMISO)-positron emission tomography (PET) at baseline, and during C1 and C5. Pre- and post-bevacizumab vascular changes were evaluated using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Molecular biomarkers were assessed using microarray analysis. A total of 70 patients were assessed for treatment efficacy. Significant decreases from baseline in tumour proliferation (FLT-PET), vascularity, and perfusion (DCE-MRI) were observed during C1 (p ≤ 0.001), independent of tumour subtype. Bevacizumab treatment did not affect hypoxic tumour status (FMISO-PET). Significant changes in the expression of 28 genes were observed after C1. Changes in vascular endothelial growth factor receptor (VEGFR)-2p levels were observed in 65 patients, with a > 20% decrease in VEGFR-2p observed in 13/65. Serial imaging techniques and molecular gene profiling identified several potentially predictive biomarkers that may predict response to neoadjuvant bevacizumab therapy in BC patients.
Collapse
Affiliation(s)
- José Manuel López-Vega
- Department of Medical Oncology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain;
| | - Isabel Álvarez
- Department of Medical Oncology, University Hospital Donostia, 20080 Donostia-San Sebastián, Spain;
| | - Antonio Antón
- Department of Medical Oncology, University Hospital Miguel Servet, 50009 Zaragoza, Spain;
| | | | - Antonio Llombart
- Department of Medical Oncology, Hospital Arnau de Vilanova, 46015 Lleida, Spain;
| | - Valentina Boni
- START Madrid CIOCC, Hospital Universitario HM Sanchinarro, 28050 Madrid, Spain;
| | | | - Josep María Martí-Climent
- Department of Medical Physics and Radiation Safety, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Luis Pina
- Department of Radiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Jesús García-Foncillas
- Translational Oncology Division, OncoHealth Institute, University Hospital “Fundación Jiménez Díaz”, Autonomous University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
20
|
Ko CC, Yeh LR, Kuo YT, Chen JH. Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomark Res 2021; 9:52. [PMID: 34215324 PMCID: PMC8252278 DOI: 10.1186/s40364-021-00306-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Response Evaluation Criteria in Solid Tumors (RECIST) is the gold standard for assessment of treatment response in solid tumors. Morphologic change of tumor size evaluated by RECIST is often correlated with survival length and has been considered as a surrogate endpoint of therapeutic efficacy. However, the detection of morphologic change alone may not be sufficient for assessing response to new anti-cancer medication in all solid tumors. During the past fifteen years, several molecular-targeted therapies and immunotherapies have emerged in cancer treatment which work by disrupting signaling pathways and inhibited cell growth. Tumor necrosis or lack of tumor progression is associated with a good therapeutic response even in the absence of tumor shrinkage. Therefore, the use of unmodified RECIST criteria to estimate morphological changes of tumor alone may not be sufficient to estimate tumor response for these new anti-cancer drugs. Several studies have reported the low reliability of RECIST in evaluating treatment response in different tumors such as hepatocellular carcinoma, lung cancer, prostate cancer, brain glioma, bone metastasis, and lymphoma. There is an increased need for new medical imaging biomarkers, considering the changes in tumor viability, metabolic activity, and attenuation, which are related to early tumor response. Promising imaging techniques, beyond RECIST, include dynamic contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI), diffusion-weight imaging (DWI), magnetic resonance spectroscopy (MRS), and 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET). This review outlines the current RECIST with their limitations and the new emerging concepts of imaging biomarkers in oncology.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ren Yeh
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeon-Hor Chen
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan. .,Tu & Yuan Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, 164 Irvine Hall, Irvine, CA, 92697 - 5020, USA.
| |
Collapse
|
21
|
The value of diffusion-weighted imaging in the diagnosis of medication-related osteonecrosis of the jaws. Oral Surg Oral Med Oral Pathol Oral Radiol 2021; 132:339-345. [PMID: 34092542 DOI: 10.1016/j.oooo.2021.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The purpose of this study was to assess apparent diffusion coefficient (ADC) values associated with medication-related osteonecrosis of the jaws (MRONJ) at each stage of the disease compared to controls. STUDY DESIGN The magnetic resonance imaging data of 38 patients in the 4 stages of MRONJ and 10 controls were analyzed. Mean ADC values of bone marrow in the controls and patients in each stage of MRONJ were calculated. The significance of differences was analyzed by using the Kruskal-Wallis test and post-hoc Mann-Whitney tests with Bonferroni adjustment. A receiver operating characteristic (ROC) curve was plotted to distinguish controls from MRONJ stage 0. P < .05 was considered statistically significant. RESULTS The mean ADC values of controls and stages 0 to 3 were significantly different, with the lowest value in the controls (P < .001). ROC analysis revealed a cutoff ADC value of 1.06 × 10-3 mm2/s to distinguish between the control group and MRONJ stage 0 patients, producing values of sensitivity, specificity, accuracy, and area under the ROC curve ranging from 0.89 to 0.94. CONCLUSIONS The ADC values of bone marrow affected by MRONJ are significantly different among controls and stages 0 to 3. ADC values may be useful for distinguishing normal from MRONJ stage 0.
Collapse
|
22
|
Pandurangi RS, Tomasetti M, Verapazham ST, Paulmurugan R, Ma C, Rajput S, Anjanappa M, Nakshatri H. A Priori Activation of Apoptosis Pathways of Tumor (AAAPT) technology: Development of targeted apoptosis initiators for cancer treatment. PLoS One 2021; 16:e0225869. [PMID: 33556062 PMCID: PMC7870153 DOI: 10.1371/journal.pone.0225869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/18/2020] [Indexed: 11/24/2022] Open
Abstract
Cancer cells develop tactics to circumvent the interventions by desensitizing themselves to interventions. Amongst many, the principle routes of desensitization include a) activation of survival pathways (e.g. NF-kB, PARP) and b) downregulation of cell death pathways (e.g. CD95/CD95L). As a result, it requires high therapeutic dose to achieve tumor regression which, in turn damages normal cells through the collateral effects. Methods are needed to sensitize the low and non-responsive resistant tumor cells including cancer stem cells (CSCs) in order to evoke a better response from the current treatments. Current treatments including chemotherapy can induce cell death only in bulk cancer cells sparing CSCs and cancer resistant cells (CRCs) which are shown to be responsible for high recurrence of disease and low patient survival. Here, we report several novel tumor targeted sensitizers derived from the natural Vitamin E analogue (AMP-001-003). The drug design is based on a novel concept "A priori activation of apoptosis pathways of tumor technology (AAAPT) which is designed to activate specific cell death pathways and inhibit survival pathways simultaneously and selectively in cancer cells sparing normal cells. Our results indicate that AMP-001-003 sensitize various types of cancer cells including MDA-MB-231 (triple negative breast cancer), PC3 (prostate cancer) and A543 (lung cancer) cells resulting in reducing the IC-50 of doxorubicin in vitro when used as a combination. At higher doses, AMP-001 acts as an anti-tumor agent on its own. The synergy between AMP-001 and doxorubicin could pave a new pathway to use AAAPT leading molecules as neoadjuvant to chemotherapy to achieve better efficacy and reduced off-target toxicity compared to the current treatments.
Collapse
Affiliation(s)
- Raghu S. Pandurangi
- Sci-Engi-Medco Solutions Inc., St Charles, Missouri, United States of America
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Section of Experimental and Occupational Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Sekar T. Verapazham
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Cynthia Ma
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sandeep Rajput
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Manjushree Anjanappa
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Harikrishna Nakshatri
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
23
|
Notohamiprodjo S, Varasteh Z, Beer AJ, Niu G, Chen X(S, Weber W, Schwaiger M. Tumor Vasculature. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
24
|
Song X, Liu C, Wang N, Huang H, He S, Gong C, Wei Y. Delivery of CRISPR/Cas systems for cancer gene therapy and immunotherapy. Adv Drug Deliv Rev 2021; 168:158-180. [PMID: 32360576 DOI: 10.1016/j.addr.2020.04.010] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/19/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
The clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems are efficient and versatile gene editing tools, which offer enormous potential to treat cancer by editing genome, transcriptome or epigenome of tumor cells and/or immune cells. A large body of works have been done with CRISPR/Cas systems for genetic modification, and 16 clinical trials were conducted to treat cancer by ex vivo or in vivo gene editing approaches. Now, promising preclinical works have begun using CRISPR/Cas systems in vivo. However, efficient and safe delivery of CRISPR/Cas systems in vivo is still a critical challenge for their clinical applications. This article summarizes delivery of CRISPR/Cas systems by physical methods, viral vectors and non-viral vectors for cancer gene therapy and immunotherapy. The prospects for the development of physical methods, viral vectors and non-viral vectors for delivery of CRISPR/Cas systems are reviewed, and promising advances in cancer treatment using CRISPR/Cas systems are discussed.
Collapse
Affiliation(s)
- Xiangrong Song
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Chao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hai Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Siyan He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
25
|
Ntellas P, Mavroeidis L, Gkoura S, Gazouli I, Amylidi AL, Papadaki A, Zarkavelis G, Mauri D, Karpathiou G, Kolettas E, Batistatou A, Pentheroudakis G. Old Player-New Tricks: Non Angiogenic Effects of the VEGF/VEGFR Pathway in Cancer. Cancers (Basel) 2020; 12:E3145. [PMID: 33121034 PMCID: PMC7692709 DOI: 10.3390/cancers12113145] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis has long been considered to facilitate and sustain cancer growth, making the introduction of anti-angiogenic agents that disrupt the vascular endothelial growth factor/receptor (VEGF/VEGFR) pathway an important milestone at the beginning of the 21st century. Originally research on VEGF signaling focused on its survival and mitogenic effects towards endothelial cells, with moderate so far success of anti-angiogenic therapy. However, VEGF can have multiple effects on additional cell types including immune and tumor cells, by directly influencing and promoting tumor cell survival, proliferation and invasion and contributing to an immunosuppressive microenvironment. In this review, we summarize the effects of the VEGF/VEGFR pathway on non-endothelial cells and the resulting implications of anti-angiogenic agents that include direct inhibition of tumor cell growth and immunostimulatory functions. Finally, we present how previously unappreciated studies on VEGF biology, that have demonstrated immunomodulatory properties and tumor regression by disrupting the VEGF/VEGFR pathway, now provide the scientific basis for new combinational treatments of immunotherapy with anti-angiogenic agents.
Collapse
Affiliation(s)
- Panagiotis Ntellas
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Leonidas Mavroeidis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Stefania Gkoura
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Ioanna Gazouli
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Anna-Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Alexandra Papadaki
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Georgia Karpathiou
- Department of Pathology, University Hospital of St-Etienne, 42055 Saint Etienne, France;
| | - Evangelos Kolettas
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Biomedical Research Division, Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology, 45115 Ioannina, Greece
| | - Anna Batistatou
- Department of Pathology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - George Pentheroudakis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| |
Collapse
|
26
|
Cappuccio S, Marinucci B, Bove V, Turco LC, Cosentino F, Scambia G, Costamagna G, Boškoski I. Rare Case of Endoscopic Treatment for Bevacizumab-Related Gastric Perforation in a Patient with Ovarian Cancer. Chemotherapy 2020; 65:54-57. [PMID: 32615577 DOI: 10.1159/000508556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/08/2020] [Indexed: 11/19/2022]
Abstract
Antiangiogenic chemotherapy is a common treatment option for patients with advanced ovarian cancer (OC) and has been proven to be effective and generally safe. Nevertheless, in rare cases, these drugs can give serious complications such as gastrointestinal perforations that can be even mortal or very difficult to treat and can heavily impact the clinical management. We present a rare case of bevacizumab-induced gastric perforation in a patient with advanced OC occurred during bevacizumab-including chemotherapy in an adjuvant setting. Surgical treatment was not possible due to the frailty of the clinical condition of the patient and the gastric perforation was successfully treated with endoscopic suturing.
Collapse
Affiliation(s)
- Serena Cappuccio
- Department of Woman's, Child's and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy,
| | - Benito Marinucci
- Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | - Vincenzo Bove
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luigi Carlo Turco
- Gynecology and Breast Care Unit, Mater Olbia Spa, Olbia, Italy.,Gynecologic Oncology, Gemelli Molise Spa, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Francesco Cosentino
- Gynecologic Oncology, Gemelli Molise Spa, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Giovanni Scambia
- Department of Woman's, Child's and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | - Guido Costamagna
- Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy.,Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ivo Boškoski
- Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy.,Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
27
|
Nguyen D, Yu J, Reinhold WC, Yang SX. Association of Independent Prognostic Factors and Treatment Modality With Survival and Recurrence Outcomes in Breast Cancer. JAMA Netw Open 2020; 3:e207213. [PMID: 32644137 PMCID: PMC7348688 DOI: 10.1001/jamanetworkopen.2020.7213] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/29/2020] [Indexed: 01/15/2023] Open
Abstract
Importance It is not well understood whether prognostic factors in breast cancer are affected by specific treatment and vary by clinical outcome type compared with untreated patients. Objective To identify independent clinical and molecular measurements associated with overall survival (OS) and recurrence-free survival (RFS) by homogeneous treatment in women with breast cancer. Design, Setting, and Participants This prognostic study included 956 patients diagnosed with invasive breast cancer from hospital centers across 4 geographical regions of the United States who participated in the accreditation program of the Commission on Cancer of the American College of Surgeons from 1985 to 1997. The duration of follow-up ranged from 1 to 282 months. The study analysis was conducted from June 10, 2019, to March 18, 2020. Main Outcomes and Measures Analysis of OS and RFS in patients who underwent chemotherapy, radiotherapy, or endocrine therapy alone compared with no systemic or locoregional therapy. Cox proportional hazards regression models were used to estimate independent performance and 95% CI of age, tumor size, number of positive nodes (nodal status), tumor grades 2 and 3, p53 status, estrogen receptor (ER) status, and ERBB2 (formerly HER2) status. Results Among 956 participants, median age was 61 (range, 25-96) years. Age (adjusted hazard ratio [AHR], 2.24; 95% CI, 1.27-3.94; P = .01) and high grade (AHR, 2.05; 95% CI, 1.09-3.86; P = .02), in addition to nodal status and tumor size, were independently associated with OS and RFS, respectively, in untreated patients. p53 status (AHR, 2.11; 95% CI, 1.07-4.18; P = .03) and ER status (AHR, 0.46; 95% CI, 0.23-0.92; P = .03) were associated with higher and lower risks of death, respectively, whereas nodal status (AHR, 1.13; 95% CI, 1.06-1.20; P < .005), high grade (AHR, 4.01; 95% CI, 1.51-10.70; P = .01), and ERBB2 positivity (AHR, 2.67; 95% CI, 1.25-5.70; P = .01) were associated with the risk of recurrence after endocrine therapy. Tumor size (AHR for OS, 2.76 [95% CI, 1.79-4.31; P < .005]; AHR for RFS, 2.27 [95% CI, 1.23-4.18; P = .01]) and ERBB2 status (AHR for OS, 5.35 [95% CI, 1.31-21.98; P = .02]; AHR for RFS, 6.05 [95% CI, 1.48-24.78; P = .01]) were independently associated with radiotherapy outcomes, and nodal status was significantly associated with chemotherapy outcomes (AHR for OS, 1.06 [95% CI, 1.02-1.09; P < .005]; AHR for RFS, 1.05 [95% CI, 1.01-1.09; P = .01]). Conclusions and Relevance In this study, independent prognostic factors were associated with specific treatment and weighted by the outcome category with reference to untreated patients within biological and clinical contexts.
Collapse
Affiliation(s)
- Dat Nguyen
- National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - John Yu
- DSC, Inc, Reston, Virginia
- currently affiliated with Bellese Technologies, LLC, Owings Mills, Maryland
| | - William C. Reinhold
- Laboratory of Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sherry X. Yang
- National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
28
|
Low-Dose Perfusion Computed Tomography for Breast Cancer to Quantify Tumor Vascularity: Correlation With Prognostic Biomarkers. Invest Radiol 2019; 54:273-281. [PMID: 30570503 DOI: 10.1097/rli.0000000000000538] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the feasibility of using low-dose perfusion computed tomography (CT) in breast cancers for quantification of tumor vascularity and to correlate perfusion indexes with prognostic biomarkers. MATERIALS AND METHODS This preliminary study was approved by our institutional review board. Signed informed consent was obtained from all 70 enrolled patients with invasive breast cancers. Low-dose perfusion CT was performed with the patient in the prone position using a spectral CT device set at 80 kVp and 30 mAs (1.30-1.40 mSv). Images were analyzed using commercial software applying the maximum slope algorithm. On CT perfusion maps, perfusion (mL/min per 100 mL), blood volume (mL/100 g), time-to-peak enhancement (second), and peak enhancement intensity (HU) were measured in the tumor, normal breast glandular tissues, and fat. Tumor grade, estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2), and Ki67 level were evaluated using histopathology. Statistically, CT perfusion indexes of the tumor and normal glandular tissues or fat were compared using the Wilcoxon signed-rank test, and CT indexes were correlated with histological characteristics using the Mann-Whitney U or Kruskal-Wallis tests. We also correlated CT indexes with magnetic resonance imaging enhancement characteristics. RESULTS In breast cancers, perfusion, blood volume, and peak enhancement intensity values were significantly higher, and time to peak was shorter than in normal glandular tissues and fat (P < 0.001). Perfusion increased significantly in high-grade, ER-, or HER2+ cancers (P < 0.05). Time to peak decreased in ER-, HER2+, and high-grade cancers or in those with high Ki67 levels (P < 0.05). Peak enhancement intensity significantly increased in high-grade cancers (P < 0.05). HER2 overexpressing cancers showed significantly higher perfusion and shorter time to peak than luminal-type cancers (P < 0.05). Perfusion increased and time to peak decreased significantly in cancers with washout enhancement patterns on magnetic resonance imaging. CONCLUSIONS Low-dose perfusion CT in the prone position is feasible to quantify tumor vascularity in breast cancers, and CT perfusion indexes are significantly correlated with prognostic biomarkers and molecular subtypes of breast cancer.
Collapse
|
29
|
Chen J, Easwaralingam N, Warrier S, Ong A, Carson EK, Mak C, Snook K, Middleton K, Parker A, Palmieri C, Spillane A, Mann GB, Lim E, Segara D. Window of opportunity treatment in breast cancer. ANZ J Surg 2019; 90:34-40. [PMID: 31770829 DOI: 10.1111/ans.15487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 11/27/2022]
Abstract
Window of opportunity therapies, which involve short-term administration of systemic therapy between cancer diagnosis and surgery, have raised significant interest in recent years as a mean of assessing the sensitivity of a patient's cancer to therapy prior to surgery. There is now compelling evidence that in patients with early stage hormone-receptor positive breast cancer, a 2-week preoperative treatment with standard hormone therapies in a preoperative window period provides important prognostic information, which in turn helps to aid decision-making regarding treatment options. Changes in short-term biomarker endpoints such as cell proliferation measured by Ki-67 can act as surrogate markers of long-term outcomes. Paired tissues obtained pre- and post-investigational treatment, without having to subject the patient to additional biopsies, can then be used to conduct translational research to investigate predictive biomarkers and pharmacodynamics. In this review, we will examine the utility and challenges of window of opportunities therapies in breast cancer in the current literature, and the current Australian and international trial landscape in this clinical space.
Collapse
Affiliation(s)
- Julia Chen
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Neshanth Easwaralingam
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Warrier
- Chris O'Brien Lifehouse and The University of Sydney, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital Institute of Academic Surgery, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew Ong
- Campbelltown Hospital and The University of Western Sydney, Sydney, New South Wales, Australia
| | - Emma-Kate Carson
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia.,Campbelltown Hospital and The University of Western Sydney, Sydney, New South Wales, Australia
| | - Cindy Mak
- Chris O'Brien Lifehouse and The University of Sydney, Sydney, New South Wales, Australia
| | - Kylie Snook
- Breast and Surgical Oncology at the Poche Centre and Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Kate Middleton
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Parker
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Carlo Palmieri
- University of Liverpool, Clatterbridge Cancer Centre, NHS Foundation Trust, and Royal Liverpool University Hospital, Liverpool, UK
| | - Andrew Spillane
- Breast and Surgical Oncology at the Poche Centre and Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, Sydney, New South Wales, Australia.,Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - G Bruce Mann
- The Royal Melbourne Hospital and The Royal Women's Hospital, Melbourne, Victoria, Australia.,Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Davendra Segara
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Inflammatory Breast Cancer: Diagnostic, Molecular and Therapeutic Considerations. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00337-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Jagannathan NR. Application of in vivo MR methods in the study of breast cancer metabolism. NMR IN BIOMEDICINE 2019; 32:e4032. [PMID: 30456917 DOI: 10.1002/nbm.4032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 08/25/2018] [Accepted: 09/26/2018] [Indexed: 06/09/2023]
Abstract
In the last two decades, various in vivo MR methodologies have been evaluated for their potential in the study of cancer metabolism. During malignant transformation, metabolic alterations occur, leading to morphological and functional changes. Among various MR methods, in vivo MRS has been extensively used in breast cancer to study the metabolism of cells, tissues or whole organs. It provides biochemical information at the metabolite level. Altered choline, phospholipid and energy metabolism has been documented using proton (1 H), phosphorus (31 P) and carbon (13 C) isotopes. Increased levels of choline-containing compounds, phosphomonoesters and phosphodiesters in breast cancer, which are indicative of altered choline and phospholipid metabolism, have been reported using in vivo, in vitro and ex vivo NMR studies. These changes are reversed on successful therapy, which depends on the treatment regimen given. Monitoring the various tumor intermediary metabolic pathways using nuclear spin hyperpolarization of 13 C-labeled substrates by dynamic nuclear polarization has also been recently reported. Furthermore, the utility of various methods such as diffusion, dynamic contrast and perfusion MRI have also been evaluated to study breast tumor metabolism. Parameters such as tumor volume, apparent diffusion coefficient, volume transfer coefficient and extracellular volume ratio are estimated. These parameters provide information on the changes in tumor microstructure, microenvironment, abnormal vasculature, permeability and grade of the tumor. Such changes seen during cancer progression are due to alterations in the tumor metabolism, leading to changes in cell architecture. Due to architectural changes, the tissue mechanical properties are altered; this can be studied using magnetic resonance elastography, which measures the elastic properties of tissues. Moreover, these structural MRI methods can be used to investigate the effect of therapy-induced changes in tumor characteristics. This review discusses the potential of various in vivo MR methodologies in the study of breast cancer metabolism.
Collapse
|
32
|
Michmerhuizen AR, Pesch AM, Moubadder L, Chandler BC, Wilder-Romans K, Cameron M, Olsen E, Thomas DG, Zhang A, Hirsh N, Ritter CL, Liu M, Nyati S, Pierce LJ, Jagsi R, Speers C. PARP1 Inhibition Radiosensitizes Models of Inflammatory Breast Cancer to Ionizing Radiation. Mol Cancer Ther 2019; 18:2063-2073. [PMID: 31413177 DOI: 10.1158/1535-7163.mct-19-0520] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/09/2019] [Accepted: 08/08/2019] [Indexed: 11/16/2022]
Abstract
Sustained locoregional control of disease is a significant issue in patients with inflammatory breast cancer (IBC), with local control rates of 80% or less at 5 years. Given the unsatisfactory outcomes for these patients, there is a clear need for intensification of local therapy, including radiation. Inhibition of the DNA repair protein PARP1 has had little efficacy as a single agent in breast cancer outside of studies restricted to patients with BRCA mutations; however, PARP1 inhibition (PARPi) may lead to the radiosensitization of aggressive tumor types. Thus, this study investigates inhibition of PARP1 as a novel and promising radiosensitization strategy in IBC. In multiple existing IBC models (SUM-149, SUM-190, MDA-IBC-3), PARPi (AZD2281-olaparib and ABT-888-veliparib) had limited single-agent efficacy (IC50 > 10 μmol/L) in proliferation assays. Despite limited single-agent efficacy, submicromolar concentrations of AZD2281 in combination with RT led to significant radiosensitization (rER 1.12-1.76). This effect was partially dependent on BRCA1 mutational status. Radiosensitization was due, at least in part, to delayed resolution of double strand DNA breaks as measured by multiple assays. Using a SUM-190 xenograft model in vivo, the combination of PARPi and RT significantly delays tumor doubling and tripling times compared with PARPi or RT alone with limited toxicity. This study demonstrates that PARPi improves the effectiveness of radiotherapy in IBC models and provides the preclinical rationale for the opening phase II randomized trial of RT ± PARPi in women with IBC (SWOG 1706, NCT03598257).
Collapse
Affiliation(s)
- Anna R Michmerhuizen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
| | - Andrea M Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Leah Moubadder
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin C Chandler
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Cancer Biology Program, University of Michigan, Ann Arbor, Michigan
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Meleah Cameron
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Eric Olsen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Dafydd G Thomas
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Amanda Zhang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Nicole Hirsh
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Cassandra L Ritter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Meilan Liu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Shyam Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Reshma Jagsi
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Center for Bioethics and Social Sciences, University of Michigan, Ann Arbor, Michigan
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan. .,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
33
|
Pokuri VK, Tomaszewski GM, Ait-Oudhia S, Groman A, Khushalani NI, Lugade AA, Thanavala Y, Ashton EA, Grande C, Fetterly GJ, Iyer R. Efficacy, Safety, and Potential Biomarkers of Sunitinib and Transarterial Chemoembolization (TACE) Combination in Advanced Hepatocellular Carcinoma (HCC): Phase II Trial. Am J Clin Oncol 2019; 41:332-338. [PMID: 27014931 DOI: 10.1097/coc.0000000000000286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To evaluate the safety/efficacy and explore biomarkers for a rationally designed combination of sunitinib and transarterial chemoembolization (TACE) in a prospective phase 2 study of advanced hepatocellular carcinoma (HCC). METHODS Inoperable HCC patients with Child-Pugh A disease received 37.5 mg sunitinib from days 1 to 7 followed by TACE on day 8. Sunitinib was resumed from days 15 to 36 followed by 2 weeks off. Patients received subsequent sunitinib cycles of 4 weeks on and 2 weeks off. Dynamic contrast-enhanced magnetic resonance imaging and circulating soluble biomarkers were assessed at baseline, day 8, day 10, and day 36. RESULTS Sixteen patients with liver only (n=10) and extrahepatic disease (n=6) were enrolled. After a median follow-up of 12.8 months, 2 partial responses, 11 stable disease, and 3 clinical deteriorations were seen for a clinical benefit rate of 81%. Median progression-free survival (PFS) was 8 months (95% CI, 4.3-9.3) and overall survival was 14.9 months (95% CI, 6.3-27.1). Eleven of 16 patients (69%) had grade 3/4 toxicities attributable to sunitinib, the most frequent being thrombocytopenia, amylase/lipase elevations, lymphopenia, and fatigue. Mean K (volume transfer constant) and viable tumor percent in consented patients decreased by 27% and 14.8%, respectively, with combination therapy. Soluble vascular endothelial growth factor receptor-2 (sVEGFR2) levels, cytokines (interleukin-8, interleukin-21), and monocytes decreased with combination therapy. Estimated sunitinib IC50 values of 15 and 10 ng/mL modulated K and AUC90. sVEGFR2 levels decreased with K and AUC90. CONCLUSIONS Encouraging progression-free survival and overall survival were seen with acceptable toxicity in our study of sunitinib and TACE combination in advanced HCC. Potential imaging and serum biomarkers showed increased benefit with combination therapy.
Collapse
Affiliation(s)
| | | | - Sihem Ait-Oudhia
- Pharmacometrics and Systems Pharmacology at Lake Nona, University of Florida, Orlando, FL
| | | | | | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Cancer Institute (RPCI), Buffalo
| | | | | | | | | | | |
Collapse
|
34
|
Stem Cells Inhibition by Bevacizumab in Combination with Neoadjuvant Chemotherapy for Breast Cancer. J Clin Med 2019; 8:jcm8050612. [PMID: 31064127 PMCID: PMC6572380 DOI: 10.3390/jcm8050612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Preclinical works have suggested cytotoxic chemotherapies may increase the number of cancer stem cells (CSC) whereas angiogenesis inhibition may decrease CSC proliferation. We developed a proof of concept clinical trial to explore bevacizumab activity on breast CSC. Breast cancer patients requiring preoperative chemotherapy were included in this open-label, randomized, prospective, multicenter phase II trial. All received FEC-docetaxel combination, and patients randomized in the experimental arm received concomitant bevacizumab. The primary endpoint was to describe ALDH1 (Aldehyde dehydrogenase 1) positive tumor cells rate before treatment and after the fourth cycle. Secondary objectives included safety, pathological complete response (pCR) rate, disease-free survival (DFS), relapse-free survival (RFS), and overall survival (OS). Seventy-five patients were included. ALDH1+ cells rate increase was below the predefined 5% threshold in both arms for the 32 patients with two time points available. Grade 3 or 4 adverse events rates were similar in both arms. A non-significant increase in pCR was observed in the bevacizumab arm (42.6% vs. 18.2%, p = 0.06), but survival was not improved (OS: p = 0.89; DFS: p = 0.45; and RFS: p = 0.68). The increase of ALDH1+ tumor cells rate after bevacizumab-based chemotherapy was less than 5%. However, as similar results were observed with chemotherapy alone, bevacizumab impact on breast CSC cells cannot be confirmed.
Collapse
|
35
|
Li D, Finley SD. The impact of tumor receptor heterogeneity on the response to anti-angiogenic cancer treatment. Integr Biol (Camb) 2019; 10:253-269. [PMID: 29623971 DOI: 10.1039/c8ib00019k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple promoters and inhibitors mediate angiogenesis, the formation of new blood vessels, and these factors represent potential targets for impeding vessel growth in tumors. Vascular endothelial growth factor (VEGF) is a potent angiogenic factor targeted in anti-angiogenic cancer therapies. In addition, thrombospondin-1 (TSP1) is a major endogenous inhibitor of angiogenesis, and TSP1 mimetics are being developed as an alternative type of anti-angiogenic agent. The combination of bevacizumab, an anti-VEGF agent, and ABT-510, a TSP1 mimetic, has been tested in clinical trials to treat advanced solid tumors. However, the patients' responses are highly variable and show disappointing outcomes. To obtain mechanistic insight into the effects of this combination anti-angiogenic therapy, we have constructed a novel whole-body systems biology model including the VEGF and TSP1 reaction networks. Using this molecular-detailed model, we investigated how the combination anti-angiogenic therapy changes the amounts of pro-angiogenic and anti-angiogenic complexes in cancer patients. We particularly focus on answering the question of how the effect of the combination therapy is influenced by tumor receptor expression, one aspect of patient-to-patient variability. Overall, this model complements the clinical administration of combination anti-angiogenic therapy, highlights the role of tumor receptor variability in the heterogeneous responses to anti-angiogenic therapy, and identifies the tumor receptor profiles that correlate with a high likelihood of a positive response to the combination therapy. Our model provides novel understanding of the VEGF-TSP1 balance in cancer patients at the systems-level and could be further used to optimize combination anti-angiogenic therapy.
Collapse
Affiliation(s)
- Ding Li
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, California 90089, USA.
| | | |
Collapse
|
36
|
Tanabe A, Kobayashi D, Maeda K, Taguchi M, Sahara H. Angiogenesis-related gene expression profile in clinical cases of canine cancer. Vet Med Sci 2019; 5:19-29. [PMID: 30265453 PMCID: PMC6376169 DOI: 10.1002/vms3.127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The balance between pro- and anti-angiogenic signalling is tightly regulated in normal tissues to maintain the functions of the vasculature. In contrast, the overproduction of angiogenic factors and enhanced angiogenesis are frequently observed in several types of tumours. Although there have been many reports on the correlation between tumour progression and angiogenesis in humans, little is known about tumour angiogenesis in canines. Hence, we attempted to clarify whether angiogenesis contributes to tumour progression in canines as well as humans. In this study, we investigated the expression of several angiogenesis-related genes, including CD34, VEGF-A, VEGFR-1, VEGFR-2, Ang-1, Ang-2, Tie1, and Tie2, in 66 canine tumour tissues and in the normal tissues surrounding the tumours by quantitative real-time PCR analysis. Our comparative analysis between canine tumour tissues and normal tissues revealed that several angiogenesis-related genes, such as vascular endothelial growth factor (VEGF) and VEGF-receptor genes, were significantly upregulated in canine tumour tissues when compared to the normal tissues. We also found that the angiopoietin (Ang)-1/Ang-2 gene expression ratio was lower in canine tumour tissues than in the normal tissues, suggesting less association between vascular endothelial cells and perivascular cells in the canine tumour tissues. Taken together, our results suggest that several angiogenesis-related genes may contribute to the malignant progression of canine tumours via tumour angiogenesis.
Collapse
Affiliation(s)
- Atsushi Tanabe
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Daisuke Kobayashi
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Koki Maeda
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Masayuki Taguchi
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Hiroeki Sahara
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| |
Collapse
|
37
|
Elsayed NM, Serya RA, Tolba MF, Ahmed M, Barakat K, Abou El Ella DA, Abouzid KA. Design, synthesis, biological evaluation and dynamics simulation of indazole derivatives with antiangiogenic and antiproliferative anticancer activity. Bioorg Chem 2019; 82:340-359. [PMID: 30428414 DOI: 10.1016/j.bioorg.2018.10.071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
|
38
|
Zhang W, Jia L, Liu DLX, Chen L, Wang Q, Song K, Nie S, Ma J, Chen X, Xiu M, Gao M, Zhao D, Zheng Y, Duan S, Dong Z, Li Z, Wang P, Fu B, Cai G, Sun X, Chen X. Serum Stem Cell Factor Level Predicts Decline in Kidney Function in Healthy Aging Adults. J Nutr Health Aging 2019; 23:813-820. [PMID: 31641730 DOI: 10.1007/s12603-019-1253-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Stem cell factor (SCF), the ligand of the c-kit receptor, actively participates in the organ reconstruction and fibrosis associated with various diseases, including kidney disease. However, it remains unclear whether SCF plays a role in kidney aging. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS In the present study, we measured the serum SCF level, estimated glomerular filtration rate (eGFR), and other biological parameters in a Chinese Han group of 892 subjects, and explored the relationship between SCF level and renal function during aging; we sought to define novel biomarkers of kidney aging. RESULTS Multiple linear regression was used to select potential indicators of decline in renal function. Only age, SCF level, and 25% maximum expiratory flow (25% MEF) were significant predictors after redundancy analysis (|r| > 0.70 and P < 0.05). Multiple linear regression showed that the relationship among eGFR, SCF level, and age could be described as follows: eGFR = 154.486 - (0.846 × age) - (0.011 × SCF level). CONCLUSIONS We found no between-gender difference in the effect of SCF on kidney aging. In conclusion, the SCF level is an ideal biomarker of renal aging and may help to predict changes in eGFR during aging.
Collapse
Affiliation(s)
- W Zhang
- Mr. Weiguang Zhang, Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Disease, National Clinic Research Center for Kidney Diseases, A28 Fuxing Road, Beijing 100853, China; Tel +86 15811088843; E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Inflammatory Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Mamouch F, Berrada N, Aoullay Z, El Khanoussi B, Errihani H. Inflammatory Breast Cancer: A Literature Review. World J Oncol 2018; 9:129-135. [PMID: 30524636 PMCID: PMC6279456 DOI: 10.14740/wjon1161] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/25/2018] [Indexed: 01/13/2023] Open
Abstract
The multidisciplinary management of inflammatory breast cancer (IBC), which is the most aggressive form of breast cancer due to its rapid proliferation, has changed over the past three decades thanks to advances in medical treatments that represent the basis of treatment, without eliminating the use of locoregional treatments including surgery and radiotherapy in the localized stages. The molecular profile determination of IBC allows the orientation towards new targeted therapeutic strategies with an impact on survival.
Collapse
Affiliation(s)
- Fouzia Mamouch
- Mohammed V University, Rabat, Morocco.,National Institute of Oncology, Rabat, Morocco
| | | | - Zineb Aoullay
- Mohammed V University, Rabat, Morocco.,National Institute of Oncology, Rabat, Morocco
| | | | - Hassan Errihani
- Mohammed V University, Rabat, Morocco.,National Institute of Oncology, Rabat, Morocco
| |
Collapse
|
41
|
Comparison of efficacy and toxicity of bevacizumab, endostar and apatinib in transgenic and human lung cancer xenograftzebrafish model. Sci Rep 2018; 8:15837. [PMID: 30367145 PMCID: PMC6203857 DOI: 10.1038/s41598-018-34030-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/05/2018] [Indexed: 12/24/2022] Open
Abstract
The poor prognosis in non-small-cell lung cancer has driven the development of novel targeted therapies. Vascular endothelial growth factor is the most potent force in mediating tumor angiogenesis, and many angiogenesis inhibitors have been developed for oncology treatment. We performed a study to characterize the efficacy, safety and tumor suppression of three lung cancer related anti-angiogenic drugs (bevacizumab, endostar and apatinib) using transgenic zebrafish embryo and human lung cancer xenotransplantation model. All three drugs demonstrated remarkable angiogenesis and tumor inhibition effect in the zebrafish model, within the nonlethal dose range. Endostar and bevacizumab showed competitive anti-tumor efficacy. The anti-tumor performance of apatinib was hamstrung by its elevated toxicity at 35 °C. The addition of pemetrexed to anti-angiogenesis therapy had no obvious additional benefit in tumors.
Collapse
|
42
|
Kannan P, Kretzschmar WW, Winter H, Warren D, Bates R, Allen PD, Syed N, Irving B, Papiez BW, Kaeppler J, Markelc B, Kinchesh P, Gilchrist S, Smart S, Schnabel JA, Maughan T, Harris AL, Muschel RJ, Partridge M, Sharma RA, Kersemans V. Functional Parameters Derived from Magnetic Resonance Imaging Reflect Vascular Morphology in Preclinical Tumors and in Human Liver Metastases. Clin Cancer Res 2018; 24:4694-4704. [PMID: 29959141 PMCID: PMC6171743 DOI: 10.1158/1078-0432.ccr-18-0033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/11/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Purpose: Tumor vessels influence the growth and response of tumors to therapy. Imaging vascular changes in vivo using dynamic contrast-enhanced MRI (DCE-MRI) has shown potential to guide clinical decision making for treatment. However, quantitative MR imaging biomarkers of vascular function have not been widely adopted, partly because their relationship to structural changes in vessels remains unclear. We aimed to elucidate the relationships between vessel function and morphology in vivo Experimental Design: Untreated preclinical tumors with different levels of vascularization were imaged sequentially using DCE-MRI and CT. Relationships between functional parameters from MR (iAUC, K trans, and BATfrac) and structural parameters from CT (vessel volume, radius, and tortuosity) were assessed using linear models. Tumors treated with anti-VEGFR2 antibody were then imaged to determine whether antiangiogenic therapy altered these relationships. Finally, functional-structural relationships were measured in 10 patients with liver metastases from colorectal cancer.Results: Functional parameters iAUC and K trans primarily reflected vessel volume in untreated preclinical tumors. The relationships varied spatially and with tumor vascularity, and were altered by antiangiogenic treatment. In human liver metastases, all three structural parameters were linearly correlated with iAUC and K trans For iAUC, structural parameters also modified each other's effect.Conclusions: Our findings suggest that MR imaging biomarkers of vascular function are linked to structural changes in tumor vessels and that antiangiogenic therapy can affect this link. Our work also demonstrates the feasibility of three-dimensional functional-structural validation of MR biomarkers in vivo to improve their biological interpretation and clinical utility. Clin Cancer Res; 24(19); 4694-704. ©2018 AACR.
Collapse
Affiliation(s)
- Pavitra Kannan
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom.
| | - Warren W Kretzschmar
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Gene Technology, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Helen Winter
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Daniel Warren
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Russell Bates
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Philip D Allen
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nigar Syed
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
- NHS, Department of Radiology, Churchill Hospital, Oxford, United Kingdom
| | - Benjamin Irving
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Bartlomiej W Papiez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Jakob Kaeppler
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Bosjtan Markelc
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Paul Kinchesh
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Stuart Gilchrist
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sean Smart
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Julia A Schnabel
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Tim Maughan
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Adrian L Harris
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ruth J Muschel
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Mike Partridge
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ricky A Sharma
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
- NIHR University College London Hospitals Biomedical Research Centre, University College London, London, United Kingdom
| | - Veerle Kersemans
- CRUK and MRC Oxford Institute for Radiation Oncology Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Lim B, Woodward WA, Wang X, Reuben JM, Ueno NT. Inflammatory breast cancer biology: the tumour microenvironment is key. Nat Rev Cancer 2018; 18:485-499. [PMID: 29703913 DOI: 10.1038/s41568-018-0010-y] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive disease that accounts for ~2-4% of all breast cancers. However, despite its low incidence rate, IBC is responsible for 7-10% of breast cancer-related mortality in Western countries. Thus, the discovery of robust biological targets and the development of more effective therapeutics in IBC are crucial. Despite major international efforts to understand IBC biology, genomic studies have not led to the discovery of distinct biological mechanisms in IBC that can be translated into novel therapeutic strategies. In this Review, we discuss these molecular profiling efforts and highlight other important aspects of IBC biology. We present the intrinsic characteristics of IBC, including stemness, metastatic potential and hormone receptor positivity; the extrinsic features of the IBC tumour microenvironment (TME), including various constituent cell types; and lastly, the communication between these intrinsic and extrinsic components. We summarize the latest perspectives on the key biological features of IBC, with particular emphasis on the TME as an important contributor to the aggressive nature of IBC. On the basis of the current understanding of IBC, we hope to develop the next generation of translational studies, which will lead to much-needed survival improvements in patients with this deadly disease.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
44
|
El Alaoui-Lasmaili K, Faivre B. Antiangiogenic therapy: Markers of response, "normalization" and resistance. Crit Rev Oncol Hematol 2018; 128:118-129. [PMID: 29958627 DOI: 10.1016/j.critrevonc.2018.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023] Open
Abstract
Currently in cancer treatment, one premise is to use antiangiogenic therapies in association with chemotherapy or radiotherapy to augment their efficacy by benefiting from the vascular "normalization" induced by antiangiogenic therapy. This concept defines the time during which the tumor blood vessels adopt normal-like morphology and functionality, i.e. the blood vessels become more mature, the perfusion augments and hypoxia decreases. To date, there is such a diversity of treatment protocols where the type of antiangiogenic to adopt, its dose and duration of administration are different, that knowing when and how to treat is problematic. In this review, we analyzed thoroughly preclinical and clinical studies that use antiangiogenic treatments to benefit from the "normalization" and showed that the effects depend on the type of antiangiogenic administrated (anti-VEGF, anti-VEGFR, Multi-Kinase Inhibitor) and on the duration of treatment. Finally, biomarkers of "normalization" and resistance that could be used in the clinic are presented.
Collapse
Affiliation(s)
| | - Béatrice Faivre
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; Université de Lorraine, Faculté de Pharmacie, Nancy, France.
| |
Collapse
|
45
|
Curigliano G. Inflammatory breast cancer and chest wall disease: The oncologist perspective. Eur J Surg Oncol 2018; 44:1142-1147. [PMID: 30032791 DOI: 10.1016/j.ejso.2018.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
Chest wall inflammatory and lymphangitic breast cancer represents a clinical spectrum and a model disease. Inflammation and the immune response have a role in the natural history of this special clinical presentation. Preclinical models and biomarker studies suggest that inflammatory breast cancer comprises a more important role for the tumour microenvironment, including immune cell infiltration and vasculogenesis, especially lympho-angiogenesis. Across this clinical continuum of the chest wall disease there is an important role of the inflammation cascade. The activation of mature dendritic cells (DCs) through toll like receptors (TLRs) or by inflammatory cytokines converts immature DCs into mature DCs that present specific antigen to T cells, thereby activating them. Maturation of DCs is accompanied by co-stimulatory molecules and secretion of inflammatory cytokines polarizing lymphocytic, macrophages and fibroblast infiltration. It is unknown whether immune cells associated to the IBC microenvironment play a role in this scenario to transiently promote epithelial to mesenchymal transition (EMT) in these cells. Immune and microenvirnment factors can induce phenotypic, morphological, and functional changes in breast cancer cells. We can hypothesize that similar inflammatory conditions in vivo may support both the rapid metastasis and tight tumor emboli that are characteristic of chest wall disease and that targeted anti-inflammatory therapy may play a role in this patient population. The current review will review biological and clinical data of this special condition.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy.
| |
Collapse
|
46
|
Yamaguchi T, Gotoh Y, Hattori H, Katsuno H, Imaizumi K. Gastrointestinal perforation during treatment with erlotinib plus bevacizumab in two patients with non-small cell lung cancer exhibiting epidermal growth factor receptor mutations: A case report. Oncol Lett 2018; 16:1046-1050. [PMID: 29963181 DOI: 10.3892/ol.2018.8708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 03/07/2017] [Indexed: 12/14/2022] Open
Abstract
A previous randomized phase II study in patients with non-small cell lung cancer (NSCLC) identified that combination treatment with erlotinib plus bevacizumab prolonged progression-free survival compared with erlotinib alone. However, combination bevacizumab and erlotinib treatment generally increased the risk of severe adverse events, including hemorrhage, thrombosis, fistula formation and gastrointestinal perforation. The present report describes two patients with NSCLC harboring epidermal growth factor receptor (EGFR) mutations, who experienced gastrointestinal perforation associated with erlotinib plus bevacizumab combination therapy. The first patient, a 67-year-old male with stage IIIB lung adenocarcinoma harboring a L858R point mutation in EGFR exon 21, received concurrent chemoradiotherapy. However, seven months later, the patient experienced a relapse and was administered erlotinib plus bevacizumab treatment. A total of two months subsequent to commencing treatment, the patient developed a perforated duodenal ulcer. The second patient, a 66-year-old male with lung adenocarcinoma harboring a deletion in EGFR exon 19 and multiple pulmonary metastases, demonstrated a partial response to erlotinib plus bevacizumab treatment. A total of seven months subsequent to starting treatment, the patient experienced lower abdominal pain, and abdominal computed tomography confirmed a diagnosis of colocutaneous fistula complicating sigmoid diverticulitis. Following repair of the perforation, both patients were restarted on erlotinib treatment alone. Gastrointestinal perforation may be a potentially severe adverse event of erlotinib plus bevacizumab combination therapy, even in the absence of tumor metastasis in the abdomen.
Collapse
Affiliation(s)
- Teppei Yamaguchi
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Yusuke Gotoh
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hidekazu Hattori
- Department of Radiology, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hidetoshi Katsuno
- Department of Surgery, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kazuyoshi Imaizumi
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
47
|
DCE-MRI for Early Prediction of Response in Hepatocellular Carcinoma after TACE and Sorafenib Therapy: A Pilot Study. J Belg Soc Radiol 2018; 102:40. [PMID: 30039052 PMCID: PMC6032464 DOI: 10.5334/jbsr.1278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: Dynamic contrast-enhanced MRI (DCE-MRI) can measure the changes in tumor blood flow, vascular permeability and interstitial and intravascular volume. The objective was to evaluate the efficacy of DCE-MRI in prediction of Barcelona Clinic Liver Cancer (BCLC) staging B or C hepatocellular carcinoma (HCC) response after treatment with transcatheter arterial chemoembolization (TACE) followed by sorafenib therapy. Methods: Sorafenib was administered four days after TACE of BCLC staging B or C HCC in 11 patients (21 lesions). DCE-MRI was performed with Gd-EOB-DTPA contrast before TACE and three and 10 days after TACE. DCE-MRI acquisitions were taken pre-contrast, hepatic arterial-dominant phase and 60, 120, 180, 240, 330, 420, 510 and 600 seconds post-contrast. Distribution volume of contrast agent (DV) and transfer constant Ktrans were calculated. Patients were grouped by mRECIST after one month or more post-TACE into responders (complete response, partial response) and non-responders (stable disease, progressive disease). Results: DV was reduced in responders at three and 10 days post-TACE (p = 0.008 and p = 0.008 respectively). DV fell in non-responders at three days (p = 0.025) but was not significantly changed from pre-TACE values after sorafenib. Sensitivity and specificity for DV 10 days post-TACE were 88% and 77% respectively. Conclusion: DV may be a useful biomarker for early prediction of therapeutic outcome in intermediate HCC.
Collapse
|
48
|
Morotti M, Dass PH, Harris AL, Lord S. Pharmacodynamic and Pharmacokinetic Markers For Anti-angiogenic Cancer Therapy: Implications for Dosing and Selection of Patients. Eur J Drug Metab Pharmacokinet 2018; 43:137-153. [PMID: 29019020 DOI: 10.1007/s13318-017-0442-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis is integral to tumour growth and invasion, and is a key target for cancer therapeutics. However, for many of the licensed indications, only a modest clinical benefit has been observed for both monoclonal antibody and small-molecule tyrosine kinase inhibitor anti-angiogenic therapy. Pre-clinical and clinical studies have attempted to evaluate circulating, imaging, genomic, pharmacokinetic, and pharmacodynamic markers that may aid both the selection of patients for treatment and define dosing. Correct dosing is likely to be critical in the context of vascular normalization to allow better delivery of concomitant anti-cancer therapy and novel imaging techniques hold much promise in the early evaluation of pharmacodynamic response to improve efficacy.
Collapse
Affiliation(s)
- Matteo Morotti
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK.
- Department of Gynaecology Oncology, University of Oxford, Oxford, UK.
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| | - Prashanth Hari Dass
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Adrian L Harris
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Simon Lord
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
49
|
Mazur J, Roy K, Kanwar JR. Recent advances in nanomedicine and survivin targeting in brain cancers. Nanomedicine (Lond) 2017; 13:105-137. [PMID: 29161215 DOI: 10.2217/nnm-2017-0286] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is a highly lethal disease, especially devastating toward both the elderly and children. This cancer has no therapeutics available to combat it, predominately due to the blood-brain barrier (BBB) preventing treatments from maintaining therapeutic levels within the brain. Recently, nanoparticle technology has entered the forefront of cancer therapy due to its ability to deliver therapeutic effects while potentially passing physiological barriers. Key nanoparticles for brain cancer treatment include glutathione targeted PEGylated liposomes, gold nanoparticles, superparamagnetic iron oxide nanoparticles and nanoparticle-albumin bound drugs, with these being discussed throughout this review. Recently, the survivin protein has gained attention as it is over-expressed in a majority of tumors. This review will briefly discuss the properties of survivin, while focusing on how both nanoparticles and survivin-targeting treatments hold potential as brain cancer therapies. This review may provide useful insight into new brain cancer treatment options, particularly survivin inhibition and nanomedicine.
Collapse
Affiliation(s)
- Jake Mazur
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Kislay Roy
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| |
Collapse
|
50
|
Cancer Metabolism and Tumor Heterogeneity: Imaging Perspectives Using MR Imaging and Spectroscopy. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:6053879. [PMID: 29114178 PMCID: PMC5654284 DOI: 10.1155/2017/6053879] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/31/2017] [Accepted: 08/27/2017] [Indexed: 12/26/2022]
Abstract
Cancer cells reprogram their metabolism to maintain viability via genetic mutations and epigenetic alterations, expressing overall dynamic heterogeneity. The complex relaxation mechanisms of nuclear spins provide unique and convertible tissue contrasts, making magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) pertinent imaging tools in both clinics and research. In this review, we summarized MR methods that visualize tumor characteristics and its metabolic phenotypes on an anatomical, microvascular, microstructural, microenvironmental, and metabolomics scale. The review will progress from the utilities of basic spin-relaxation contrasts in cancer imaging to more advanced imaging methods that measure tumor-distinctive parameters such as perfusion, water diffusion, magnetic susceptibility, oxygenation, acidosis, redox state, and cell death. Analytical methods to assess tumor heterogeneity are also reviewed in brief. Although the clinical utility of tumor heterogeneity from imaging is debatable, the quantification of tumor heterogeneity using functional and metabolic MR images with development of robust analytical methods and improved MR methods may offer more critical roles of tumor heterogeneity data in clinics. MRI/MRS can also provide insightful information on pharmacometabolomics, biomarker discovery, disease diagnosis and prognosis, and treatment response. With these future directions in mind, we anticipate the widespread utilization of these MR-based techniques in studying in vivo cancer biology to better address significant clinical needs.
Collapse
|